1
|
Zhou W, Munoz JR, Henry Ho HY, Hanamura K, Dalva MB. Specific neuroblast-derived signals control both cell migration and fate in the rostral migratory stream. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.02.18.638163. [PMID: 40027825 PMCID: PMC11870606 DOI: 10.1101/2025.02.18.638163] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 03/05/2025]
Abstract
Functional neuronal circuits require neuroblasts migrate to appropriate locations and then differentiate into neuronal subtypes. However, it remains unknown how neuroblasts in the subventricular zone (SVZ) are guided through the rostral migratory stream (RMS) to the olfactory bulb (OB). Here we define EphB2 as a neuroblast-derived cue that controls migration along the RMS and helps to determine cell fate. Within the RMS, EphB2 is expressed selectively in, kinase-active in, and required for the migration of neuroblasts. As neuroblasts enter the OB and differentiate, EphB kinase activity is down-regulated, and in the granule cell layer (GCL), EphB2 expression is down-regulated. Blocking EphB kinase activity or knocking down EphB2 results in defects in migration and premature cellular differentiation in the RMS. Unexpectedly, premature loss of EphB2 expression causes neuroblasts to stop migrating and differentiate into astrocyte-like cells. Thus, EphB2 kinase activity and expression are linked to migration and specification of neuroblast fate.
Collapse
Affiliation(s)
- Wei Zhou
- Department of Neuroscience and the Jefferson Synaptic Biology Center, Thomas Jefferson University, 900 Walnut Street, Philadelphia, PA 19107, USA
| | - James R. Munoz
- Present address: Department of Psychology and Neuroscience, Nova Southeastern University, 3301 College Avenue, Fort Lauderdale, FL 33314, USA
| | - Hsin-Yi Henry Ho
- Department of Cell Biology and Human Anatomy, UC Davis School of Medicine, One Shields Avenue, Davis, CA 95616
| | - Kenji Hanamura
- Department of Neuroscience and the Jefferson Synaptic Biology Center, Thomas Jefferson University, 900 Walnut Street, Philadelphia, PA 19107, USA
- Present address: Faculty of Medical Technology, Department of Radiological Technology, Niigata University of Health and Welfare, 1398 Shimami-cho, Kita-ku, Niigata-City, 950-3198, Japan
| | - Matthew B. Dalva
- Department of Neuroscience and the Jefferson Synaptic Biology Center, Thomas Jefferson University, 900 Walnut Street, Philadelphia, PA 19107, USA
- Present Address: Tulane Brain Institute, Tulane University, 201 Flower Hall, 6823 St. Charles Avenue, New Orleans, LA 70118
| |
Collapse
|
2
|
Yang Y, Wang H, Liu Y, Zhai S, Liu H, He D. A novel codominant plumage color pattern of white breast patches in WugangTong geese was controlled by EDNRB2. Poult Sci 2024; 103:104324. [PMID: 39353325 PMCID: PMC11472611 DOI: 10.1016/j.psj.2024.104324] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2024] [Revised: 09/05/2024] [Accepted: 09/07/2024] [Indexed: 10/04/2024] Open
Abstract
Two basic plumage color patterns are observed in adult geese: solid grey (G) or colorless white (W). However, a Chinese indigenous breed, the Wugangtong goose (WGT), continues to be subject to selective breeding efforts as it displays segregation of plumage colors, including G, W, and a novel color pattern designated Wb (G with white breast circles). The underlying genetic mechanisms responsible for the Wb phenotype are yet to be determined. The current study employed the population differentiation index (FST) to analyze 90 geese exhibiting diverse plumage colors, identifying the fifth intron of EDNRB2 as a particularly noteworthy region with the highest FST values. Sanger sequencing of the region surrounding the EDNRB2 gene identified a 14-bp insertion within exon 3 as the causal mutation. The heterozygosity of this 14-bp insertion and wild-type alleles was completely associated with the Wb phenotype, thereby substantiating the codominant nature of the G and W phenotypes. An inter-species corroborated this finding cross between the graylag (no 14-bp insertion) and the swan goose (homozygous for the 14-bp insertion) breeds, as hybrids from this cross exhibited the Wb phenotype. Transcriptomes from white breast patches and gray dorsal skins of 4 Wb geese were compared. A significant downregulation of genes involved in melanin synthesis and melanocyte development was observed, including EDRNB2 and MLANA. The downregulation of MLANA indicated that the mutated EDNRB2 resulted in melanocyte loss in specific body regions, as MLANA is a marker gene for melanocytes. The findings were corroborated by melanin staining using the Mansson-Fontana method, which revealed no melanin particles deposited in the white breast patches. In summary, the gray plumage color was codominant to the white color in WGT geese, and plumage color variations were controlled by EDNRB2. The findings of our study offer valuable and practical guidance for the purification of plumage colors among WGT, whether through traditional phenotype selection or molecular breeding methods.
Collapse
Affiliation(s)
- Yunzhou Yang
- Institute of Animal Husbandry & Veterinary Science, Shanghai Academy of Agricultural Sciences, Shanghai, 201106, PR China
| | - Huiying Wang
- Institute of Animal Husbandry & Veterinary Science, Shanghai Academy of Agricultural Sciences, Shanghai, 201106, PR China
| | - Yi Liu
- Institute of Animal Husbandry & Veterinary Science, Shanghai Academy of Agricultural Sciences, Shanghai, 201106, PR China
| | - Shaojia Zhai
- Institute of Animal Husbandry & Veterinary Science, Shanghai Academy of Agricultural Sciences, Shanghai, 201106, PR China; College of Animal Science and Technology, Yunnan Agricultural University, Kunming, 650500, PR China
| | - Haodong Liu
- Institute of Animal Husbandry & Veterinary Science, Shanghai Academy of Agricultural Sciences, Shanghai, 201106, PR China; College of Animal Science and Technology, Shanghai Ocean University, Shanghai, 201306, PR China
| | - Daqian He
- Institute of Animal Husbandry & Veterinary Science, Shanghai Academy of Agricultural Sciences, Shanghai, 201106, PR China.
| |
Collapse
|
3
|
Jaszczak RG, Zussman JW, Wagner DE, Laird DJ. Comprehensive profiling of migratory primordial germ cells reveals niche-specific differences in non-canonical Wnt and Nodal-Lefty signaling in anterior vs posterior migrants. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.08.29.610420. [PMID: 39257761 PMCID: PMC11383659 DOI: 10.1101/2024.08.29.610420] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/12/2024]
Abstract
Mammalian primordial germ cells (PGCs) migrate asynchronously through the embryonic hindgut and dorsal mesentery to reach the gonads. We previously found that interaction with different somatic niches regulates PGC proliferation along the migration route. To characterize transcriptional heterogeneity of migrating PGCs and their niches, we performed single-cell RNA sequencing of 13,262 mouse PGCs and 7,868 surrounding somatic cells during migration (E9.5, E10.5, E11.5) and in anterior versus posterior locations to enrich for leading and lagging migrants. Analysis of PGCs by position revealed dynamic gene expression changes between faster or earlier migrants in the anterior and slower or later migrants in the posterior at E9.5; these differences include migration-associated actin polymerization machinery and epigenetic reprogramming-associated genes. We furthermore identified changes in signaling with various somatic niches, notably strengthened interactions with hindgut epithelium via non-canonical WNT (ncWNT) in posterior PGCs compared to anterior. Reanalysis of a previously published dataset suggests that ncWNT signaling from the hindgut epithelium to early migratory PGCs is conserved in humans. Trajectory inference methods identified putative differentiation trajectories linking cell states across timepoints and from posterior to anterior in our mouse dataset. At E9.5, we mainly observed differences in cell adhesion and actin cytoskeletal dynamics between E9.5 posterior and anterior migrants. At E10.5, we observed divergent gene expression patterns between putative differentiation trajectories from posterior to anterior including Nodal signaling response genes Lefty1, Lefty2, and Pycr2 and reprogramming factors Dnmt1, Prc1, and Tet1. At E10.5, we experimentally validated anterior migrant-specific Lefty1/2 upregulation via whole-mount immunofluorescence staining for LEFTY1/2 proteins, suggesting that elevated autocrine Nodal signaling accompanies the late stages of PGC migration. Together, this positional and temporal atlas of mouse PGCs supports the idea that niche interactions along the migratory route elicit changes in proliferation, actin dynamics, pluripotency, and epigenetic reprogramming.
Collapse
Affiliation(s)
| | | | - Daniel E. Wagner
- Eli and Edythe Broad Center for Regeneration Medicine and Stem Cell Research and Department of Obstetrics, Gynecology and Reproductive Science, UCSF, San Francisco, CA 94143 USA
| | - Diana J. Laird
- Eli and Edythe Broad Center for Regeneration Medicine and Stem Cell Research and Department of Obstetrics, Gynecology and Reproductive Science, UCSF, San Francisco, CA 94143 USA
| |
Collapse
|
4
|
Corallo D, Dalla Vecchia M, Lazic D, Taschner-Mandl S, Biffi A, Aveic S. The molecular basis of tumor metastasis and current approaches to decode targeted migration-promoting events in pediatric neuroblastoma. Biochem Pharmacol 2023; 215:115696. [PMID: 37481138 DOI: 10.1016/j.bcp.2023.115696] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2023] [Revised: 07/12/2023] [Accepted: 07/12/2023] [Indexed: 07/24/2023]
Abstract
Cell motility is a crucial biological process that plays a critical role in the development of multicellular organisms and is essential for tissue formation and regeneration. However, uncontrolled cell motility can lead to the development of various diseases, including neoplasms. In this review, we discuss recent advances in the discovery of regulatory mechanisms underlying the metastatic spread of neuroblastoma, a solid pediatric tumor that originates in the embryonic migratory cells of the neural crest. The highly motile phenotype of metastatic neuroblastoma cells requires targeting of intracellular and extracellular processes, that, if affected, would be helpful for the treatment of high-risk patients with neuroblastoma, for whom current therapies remain inadequate. Development of new potentially migration-inhibiting compounds and standardized preclinical approaches for the selection of anti-metastatic drugs in neuroblastoma will also be discussed.
Collapse
Affiliation(s)
- Diana Corallo
- Laboratory of Target Discovery and Biology of Neuroblastoma, Istituto di Ricerca Pediatrica (IRP), Fondazione Città della Speranza, 35127 Padova, Italy
| | - Marco Dalla Vecchia
- Laboratory of Target Discovery and Biology of Neuroblastoma, Istituto di Ricerca Pediatrica (IRP), Fondazione Città della Speranza, 35127 Padova, Italy
| | - Daria Lazic
- St. Anna Children's Cancer Research Institute, CCRI, Zimmermannplatz 10, 1090, Vienna, Austria
| | - Sabine Taschner-Mandl
- St. Anna Children's Cancer Research Institute, CCRI, Zimmermannplatz 10, 1090, Vienna, Austria
| | - Alessandra Biffi
- Pediatric Hematology, Oncology and Stem Cell Transplant Division, Woman's and Child Health Department, University of Padova, 35121 Padova, Italy
| | - Sanja Aveic
- Laboratory of Target Discovery and Biology of Neuroblastoma, Istituto di Ricerca Pediatrica (IRP), Fondazione Città della Speranza, 35127 Padova, Italy.
| |
Collapse
|
5
|
Nekooie Marnany N, Fodil R, Féréol S, Dady A, Depp M, Relaix F, Motterlini R, Foresti R, Duband JL, Dufour S. Glucose oxidation drives trunk neural crest cell development and fate. J Cell Sci 2023; 136:jcs260607. [PMID: 37589341 DOI: 10.1242/jcs.260607] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2022] [Accepted: 06/30/2023] [Indexed: 08/18/2023] Open
Abstract
Bioenergetic metabolism is a key regulator of cellular function and signaling, but how it can instruct the behavior of cells and their fate during embryonic development remains largely unknown. Here, we investigated the role of glucose metabolism in the development of avian trunk neural crest cells (NCCs), a migratory stem cell population of the vertebrate embryo. We uncovered that trunk NCCs display glucose oxidation as a prominent metabolic phenotype, in contrast to what is seen for cranial NCCs, which instead rely on aerobic glycolysis. In addition, only one pathway downstream of glucose uptake is not sufficient for trunk NCC development. Indeed, glycolysis, mitochondrial respiration and the pentose phosphate pathway are all mobilized and integrated for the coordinated execution of diverse cellular programs, epithelial-to-mesenchymal transition, adhesion, locomotion, proliferation and differentiation, through regulation of specific gene expression. In the absence of glucose, the OXPHOS pathway fueled by pyruvate failed to promote trunk NCC adaptation to environmental stiffness, stemness maintenance and fate-decision making. These findings highlight the need for trunk NCCs to make the most of the glucose pathway potential to meet the high metabolic demands appropriate for their development.
Collapse
Affiliation(s)
| | - Redouane Fodil
- Université Paris-Est Créteil, INSERM, IMRB, 94010 Créteil, France
| | - Sophie Féréol
- Université Paris-Est Créteil, INSERM, IMRB, 94010 Créteil, France
| | - Alwyn Dady
- Laboratoire Gly-CRRET, Université Paris-Est Créteil, 94000 Créteil, France
| | - Marine Depp
- Université Paris-Est Créteil, INSERM, IMRB, 94010 Créteil, France
| | - Frederic Relaix
- Université Paris-Est Créteil, INSERM, IMRB, 94010 Créteil, France
| | | | - Roberta Foresti
- Université Paris-Est Créteil, INSERM, IMRB, 94010 Créteil, France
| | - Jean-Loup Duband
- Université Paris-Est Créteil, INSERM, IMRB, 94010 Créteil, France
| | - Sylvie Dufour
- Université Paris-Est Créteil, INSERM, IMRB, 94010 Créteil, France
| |
Collapse
|
6
|
Lencer E, Rains A, Binne E, Prekeris R, Artinger KB. Mutations in cdon and boc affect trunk neural crest cell migration and slow-twitch muscle development in zebrafish. Development 2023; 150:dev201304. [PMID: 37390228 PMCID: PMC10357035 DOI: 10.1242/dev.201304] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2022] [Accepted: 06/22/2023] [Indexed: 07/02/2023]
Abstract
The transmembrane proteins cdon and boc are implicated in regulating hedgehog signaling during vertebrate development. Recent work showing roles for these genes in axon guidance and neural crest cell migration suggest that cdon and boc may play additional functions in regulating directed cell movements. We use newly generated and existing mutants to investigate a role for cdon and boc in zebrafish neural crest cell migration. We find that single mutant embryos exhibit normal neural crest phenotypes, but that neural crest migration is strikingly disrupted in double cdon;boc mutant embryos. We further show that this migration phenotype is associated with defects in the differentiation of slow-twitch muscle cells, and the loss of a Col1a1a-containing extracellular matrix, suggesting that neural crest defects may be a secondary consequence to defects in mesoderm development. Combined, our data add to a growing literature showing that cdon and boc act synergistically to promote hedgehog signaling during vertebrate development, and suggest that the zebrafish can be used to study the function of hedgehog receptor paralogs.
Collapse
Affiliation(s)
- Ezra Lencer
- Department of Cell and Developmental Biology, University of Colorado, Anschutz Medical Campus Aurora, CO 80045, USA
- Department of Craniofacial Biology, University of Colorado, Anschutz Medical Campus Aurora, CO 80045, USA
| | - Addison Rains
- Department of Craniofacial Biology, University of Colorado, Anschutz Medical Campus Aurora, CO 80045, USA
- Cell Biology, Stem Cells and Development Graduate Program, University of Colorado, Anschutz Medical Campus Aurora, CO 80045, USA
| | - Erin Binne
- Department of Craniofacial Biology, University of Colorado, Anschutz Medical Campus Aurora, CO 80045, USA
| | - Rytis Prekeris
- Department of Cell and Developmental Biology, University of Colorado, Anschutz Medical Campus Aurora, CO 80045, USA
| | - Kristin B. Artinger
- Department of Craniofacial Biology, University of Colorado, Anschutz Medical Campus Aurora, CO 80045, USA
- Department of Diagnostic and Biological Sciences, University of Minnesota School of Dentistry, Minneapolis, MN 55455, USA
| |
Collapse
|
7
|
Lawson-Keister E, Manning ML. Collective chemotaxis in a Voronoi model for confluent clusters. Biophys J 2022; 121:4624-4634. [PMID: 36299235 PMCID: PMC9748360 DOI: 10.1016/j.bpj.2022.10.029] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2021] [Revised: 06/23/2022] [Accepted: 10/19/2022] [Indexed: 12/13/2022] Open
Abstract
Collective chemotaxis, where single cells cannot climb a biochemical signaling gradient but clusters of cells can, has been observed in different biological contexts, including confluent tissues where there are no gaps or overlaps between cells. Although particle-based models have been developed that predict important features of collective chemotaxis, the mechanisms in those models depend on particle overlaps, and so it remains unclear if they can explain behavior in confluent systems. Here, we develop an open-source code that couples a two-dimensional Voronoi simulation for confluent cell mechanics to a dynamic chemical signal that can diffuse, advect, and/or degrade and use the code to study potential mechanisms for collective chemotaxis in cellular monolayers. We first study the impact of advection on collective chemotaxis and delineate a regime where advective terms are important. Next, we investigate two possible chemotactic mechanisms, contact inhibition of locomotion and heterotypic interfacial tension, and demonstrate that both can drive collective chemotaxis in certain parameter regimes. We further demonstrate that the scaling behavior of cluster motion is well captured by simple analytic theories.
Collapse
Affiliation(s)
- E Lawson-Keister
- Department of Physics and BioInspired Syracuse, Syracuse University, Syracuse, New York
| | - M L Manning
- Department of Physics and BioInspired Syracuse, Syracuse University, Syracuse, New York.
| |
Collapse
|
8
|
Howard AGA, Uribe RA. Hox proteins as regulators of extracellular matrix interactions during neural crest migration. Differentiation 2022; 128:26-32. [PMID: 36228422 PMCID: PMC10802151 DOI: 10.1016/j.diff.2022.09.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2022] [Revised: 09/23/2022] [Accepted: 09/25/2022] [Indexed: 01/19/2023]
Abstract
Emerging during embryogenesis, the neural crest are a migratory, transient population of multipotent stem cell that differentiates into various cell types in vertebrates. Neural crest cells arise along the anterior-posterior extent of the neural tube, delaminate and migrate along routes to their final destinations. The factors that orchestrate how neural crest cells undergo delamination and their subsequent sustained migration is not fully understood. This review provides a primer about neural crest epithelial-to-mesenchymal transition (EMT), with a special emphasis on the role of the Extracellular matrix (ECM), cellular effector proteins of EMT, and subsequent migration. We also summarize published findings that link the expression of Hox transcription factors to EMT and ECM modification, thereby implicating Hox factors in regulation of EMT and ECM remodeling during neural crest cell ontogenesis.
Collapse
Affiliation(s)
- Aubrey G A Howard
- BioSciences Department, Rice University, Houston, TX, 77005, USA; Biochemistry and Cell Biology Program, Rice University, Houston, TX, 77005, USA
| | - Rosa A Uribe
- BioSciences Department, Rice University, Houston, TX, 77005, USA; Biochemistry and Cell Biology Program, Rice University, Houston, TX, 77005, USA.
| |
Collapse
|
9
|
Wada Y, Tsukatani H, Kuroda C, Miyazaki Y, Otoshi M, Kobayashi I. Jagged 2b induces intercellular signaling within somites to establish hematopoietic stem cell fate in zebrafish. Development 2022; 149:274970. [DOI: 10.1242/dev.200339] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2021] [Accepted: 03/17/2022] [Indexed: 11/20/2022]
Abstract
ABSTRACT
During development, the somites play a key role in the specification of hematopoietic stem cells (HSCs). In zebrafish, the somitic Notch ligands Delta-c (Dlc) and Dld, both of which are regulated by Wnt16, directly instruct HSC fate in a shared vascular precursor. However, it remains unclear how this signaling cascade is spatially and temporally regulated within somites. Here, we show in zebrafish that an additional somitic Notch ligand, Jagged 2b (Jag2b), induces intercellular signaling to drive wnt16 expression. Jag2b activated Notch signaling in segmented somites at the early stage of somitogenesis. Loss of jag2b led to a reduction in the expression of wnt16 in the somites and an HSC marker, runx1, in the dorsal aorta, whereas overexpression of jag2b increased both. However, Notch-activated cells were adjacent to, but did not overlap with, wnt16-expressing cells within the somites, suggesting that an additional signaling molecule mediates this intercellular signal transduction. We uncover that Jag2b-driven Notch signaling induces efna1b expression, which regulates wnt16 expression in neighboring somitic cells. Collectively, we provide evidence for previously unidentified spatiotemporal regulatory mechanisms of HSC specification by somites.
Collapse
Affiliation(s)
- Yukino Wada
- Division of Life Sciences, Graduate School of Natural Science and Technology, Kanazawa University, Ishikawa 920-1192, Japan
| | - Hikaru Tsukatani
- Faculty of Natural System, Institute of Science and Engineering, Kanazawa University, Ishikawa 920-1192, Japan
| | - Chihiro Kuroda
- Faculty of Natural System, Institute of Science and Engineering, Kanazawa University, Ishikawa 920-1192, Japan
| | - Yurika Miyazaki
- Faculty of Natural System, Institute of Science and Engineering, Kanazawa University, Ishikawa 920-1192, Japan
| | - Miku Otoshi
- Faculty of Biological Science and Technology, Institute of Science and Engineering, Kanazawa University, Ishikawa 920-1192, Japan
| | - Isao Kobayashi
- Faculty of Biological Science and Technology, Institute of Science and Engineering, Kanazawa University, Ishikawa 920-1192, Japan
| |
Collapse
|
10
|
Cai ZR, McCuaig C, Hatami A, Rivière JB, Marcoux D. A novel pathogenic RHOA variant in a patient with patterned cutaneous hypopigmentation associated with extracutaneous findings. Pediatr Dermatol 2022; 39:281-287. [PMID: 35178721 PMCID: PMC9305257 DOI: 10.1111/pde.14923] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/20/2021] [Revised: 01/04/2022] [Accepted: 01/08/2022] [Indexed: 01/12/2023]
Abstract
RHOA-related neuroectodermal syndrome is characterised by linear skin hypopigmentation along Blaschko's lines associated with alopecia, leukoencephalopathy, facial and limb hypoplasia, and ocular, dental, and acral anomalies. Herein, we report a patient with patterned cutaneous hypopigmentation with a similar phenotype due to a novel postzygotic RHOA variant (c.210G>T; p.Arg70Ser). This illustrates that the complexity of the orchestration of morphogenesis and organogenesis can be affected by different variants in the same gene.
Collapse
Affiliation(s)
- Zhuo Ran Cai
- Division of Pediatric Dermatology, Department of Pediatrics, Sainte-Justine University Hospital Centre, University of Montreal, Montreal, Canada
| | - Catherine McCuaig
- Division of Pediatric Dermatology, Department of Pediatrics, Sainte-Justine University Hospital Centre, University of Montreal, Montreal, Canada
| | - Afshin Hatami
- Division of Pediatric Dermatology, Department of Pediatrics, Sainte-Justine University Hospital Centre, University of Montreal, Montreal, Canada
| | - Jean-Baptiste Rivière
- Department of Human Genetics, Faculty of Medicine, McGill University, Montreal, Canada.,Child Health and Human Development Program, Research Institute of the McGill University Health Centre, Montreal, Canada
| | - Danielle Marcoux
- Division of Pediatric Dermatology, Department of Pediatrics, Sainte-Justine University Hospital Centre, University of Montreal, Montreal, Canada
| |
Collapse
|
11
|
Bereza S, Yong R, Gronthos S, Arthur A, Ranjitkar S, Anderson PJ. Craniomaxillofacial morphology in a murine model of ephrinB1 conditional deletion in osteoprogenitor cells. Arch Oral Biol 2022; 137:105389. [DOI: 10.1016/j.archoralbio.2022.105389] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2022] [Revised: 02/26/2022] [Accepted: 02/28/2022] [Indexed: 11/02/2022]
|
12
|
From Bipotent Neuromesodermal Progenitors to Neural-Mesodermal Interactions during Embryonic Development. Int J Mol Sci 2021; 22:ijms22179141. [PMID: 34502050 PMCID: PMC8431582 DOI: 10.3390/ijms22179141] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2021] [Revised: 08/22/2021] [Accepted: 08/23/2021] [Indexed: 11/17/2022] Open
Abstract
To ensure the formation of a properly patterned embryo, multiple processes must operate harmoniously at sequential phases of development. This is implemented by mutual interactions between cells and tissues that together regulate the segregation and specification of cells, their growth and morphogenesis. The formation of the spinal cord and paraxial mesoderm derivatives exquisitely illustrate these processes. Following early gastrulation, while the vertebrate body elongates, a population of bipotent neuromesodermal progenitors resident in the posterior region of the embryo generate both neural and mesodermal lineages. At later stages, the somitic mesoderm regulates aspects of neural patterning and differentiation of both central and peripheral neural progenitors. Reciprocally, neural precursors influence the paraxial mesoderm to regulate somite-derived myogenesis and additional processes by distinct mechanisms. Central to this crosstalk is the activity of the axial notochord, which, via sonic hedgehog signaling, plays pivotal roles in neural, skeletal muscle and cartilage ontogeny. Here, we discuss the cellular and molecular basis underlying this complex developmental plan, with a focus on the logic of sonic hedgehog activities in the coordination of the neural-mesodermal axis.
Collapse
|
13
|
Muppirala AN, Limbach LE, Bradford EF, Petersen SC. Schwann cell development: From neural crest to myelin sheath. WILEY INTERDISCIPLINARY REVIEWS-DEVELOPMENTAL BIOLOGY 2020; 10:e398. [PMID: 33145925 DOI: 10.1002/wdev.398] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/09/2020] [Revised: 10/06/2020] [Accepted: 10/07/2020] [Indexed: 12/16/2022]
Abstract
Vertebrate nervous system function requires glial cells, including myelinating glia that insulate axons and provide trophic support that allows for efficient signal propagation by neurons. In vertebrate peripheral nervous systems, neural crest-derived glial cells known as Schwann cells (SCs) generate myelin by encompassing and iteratively wrapping membrane around single axon segments. SC gliogenesis and neurogenesis are intimately linked and governed by a complex molecular environment that shapes their developmental trajectory. Changes in this external milieu drive developing SCs through a series of distinct morphological and transcriptional stages from the neural crest to a variety of glial derivatives, including the myelinating sublineage. Cues originate from the extracellular matrix, adjacent axons, and the developing SC basal lamina to trigger intracellular signaling cascades and gene expression changes that specify stages and transitions in SC development. Here, we integrate the findings from in vitro neuron-glia co-culture experiments with in vivo studies investigating SC development, particularly in zebrafish and mouse, to highlight critical factors that specify SC fate. Ultimately, we connect classic biochemical and mutant studies with modern genetic and visualization tools that have elucidated the dynamics of SC development. This article is categorized under: Signaling Pathways > Cell Fate Signaling Nervous System Development > Vertebrates: Regional Development.
Collapse
Affiliation(s)
- Anoohya N Muppirala
- Program in Neuroscience, Harvard Medical School, Boston, Massachusetts, USA.,Department of Neuroscience, Kenyon College, Gambier, Ohio, USA
| | | | | | - Sarah C Petersen
- Department of Neuroscience, Kenyon College, Gambier, Ohio, USA.,Department of Biology, Kenyon College, Gambier, Ohio, USA
| |
Collapse
|
14
|
Garcia-Diaz B, Bachelin C, Coulpier F, Gerschenfeld G, Deboux C, Zujovic V, Charnay P, Topilko P, Baron-Van Evercooren A. Blood vessels guide Schwann cell migration in the adult demyelinated CNS through Eph/ephrin signaling. Acta Neuropathol 2019; 138:457-476. [PMID: 31011859 PMCID: PMC6689289 DOI: 10.1007/s00401-019-02011-1] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2019] [Revised: 03/27/2019] [Accepted: 04/07/2019] [Indexed: 12/30/2022]
Abstract
Schwann cells (SC) enter the central nervous system (CNS) in pathophysiological conditions. However, how SC invade the CNS to remyelinate central axons remains undetermined. We studied SC migratory behavior ex vivo and in vivo after exogenous transplantation in the demyelinated spinal cord. The data highlight for the first time that SC migrate preferentially along blood vessels in perivascular extracellular matrix (ECM), avoiding CNS myelin. We demonstrate in vitro and in vivo that this migration route occurs by virtue of a dual mode of action of Eph/ephrin signaling. Indeed, EphrinB3, enriched in myelin, interacts with SC Eph receptors, to drive SC away from CNS myelin, and triggers their preferential adhesion to ECM components, such as fibronectin via integrinβ1 interactions. This complex interplay enhances SC migration along the blood vessel network and together with lesion-induced vascular remodeling facilitates their timely invasion of the lesion site. These novel findings elucidate the mechanism by which SC invade and contribute to spinal cord repair.
Collapse
|
15
|
Abstract
Neural crest cells are a transient embryonic cell population that migrate collectively to various locations throughout the embryo to contribute a number of cell types to several organs. After induction, the neural crest delaminates and undergoes an epithelial-to-mesenchymal transition before migrating through intricate yet characteristic paths. The neural crest exhibits a variety of migratory behaviors ranging from sheet-like mass migration in the cephalic regions to chain migration in the trunk. During their journey, neural crest cells rely on a range of signals both from their environment and within the migrating population for navigating through the embryo as a collective. Here we review these interactions and mechanisms, including chemotactic cues of neural crest cells' migration.
Collapse
Affiliation(s)
- András Szabó
- Department of Cell and Developmental Biology, University College London, London WC1E 6BT, United Kingdom;
| | - Roberto Mayor
- Department of Cell and Developmental Biology, University College London, London WC1E 6BT, United Kingdom;
| |
Collapse
|
16
|
Logan SM, Benson MD. Medial epithelial seam cell migration during palatal fusion. J Cell Physiol 2019; 235:1417-1424. [PMID: 31264714 DOI: 10.1002/jcp.29061] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2018] [Accepted: 06/13/2019] [Indexed: 12/13/2022]
Abstract
The mammalian secondary palate forms from two shelves of mesenchyme sheathed in a single-layered epithelium. These shelves meet during embryogenesis to form the midline epithelial seam (MES). Failure of MES degradation prevents mesenchymal confluence and results in a cleft palate. Previous studies indicated that MES cells undergo features of epithelial-to-mesenchymal transition (EMT) and may become migratory as part of the fusion mechanism. To detect MES cell movement over the course of fusion, we imaged the midline of fusing embryonic ephrin-B2/GFP mouse palates in real time using two-photon microscopy. These mice express an ephrin-B2-driven green fluorescent protein (GFP) that labels the palatal epithelium nuclei and persists in those cells through the time window necessary for fusion. We observed collective migration of MES cells toward the oral surface of the palatal shelf over 48 hr of imaging, and we confirmed histologically that the imaged palates had fused by the end of the imaged period. We previously reported that ephrin reverse signaling in the MES is required for palatal fusion. We therefore added recombinant EphA4/Fc protein to block this signaling in imaged palates. The blockage inhibited fusion, as expected, but did not change the observed migration of GFP-labeled cells. Thus, we uncoupled migration and fusion. Our data reveal that palatal MES cells undergo a collective, unidirectional movement during palatal fusion and that ephrin reverse signaling, though required for fusion, controls aspects of the fusion mechanism independent of migration.
Collapse
Affiliation(s)
- Shaun M Logan
- Department of Biomedical Sciences, Texas A&M University College of Dentistry, Dallas, Texas
| | - M Douglas Benson
- Department of Biomedical Sciences, Texas A&M University College of Dentistry, Dallas, Texas
| |
Collapse
|
17
|
Lukacs M, Roberts T, Chatuverdi P, Stottmann RW. Glycosylphosphatidylinositol biosynthesis and remodeling are required for neural tube closure, heart development, and cranial neural crest cell survival. eLife 2019; 8:45248. [PMID: 31232685 PMCID: PMC6611694 DOI: 10.7554/elife.45248] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2019] [Accepted: 06/05/2019] [Indexed: 01/10/2023] Open
Abstract
Glycosylphosphatidylinositol (GPI) anchors attach nearly 150 proteins to the cell membrane. Patients with pathogenic variants in GPI biosynthesis genes develop diverse phenotypes including seizures, dysmorphic facial features and cleft palate through an unknown mechanism. We identified a novel mouse mutant (cleft lip/palate, edema and exencephaly; Clpex) with a hypo-morphic mutation in Post-Glycophosphatidylinositol Attachment to Proteins-2 (Pgap2), a component of the GPI biosynthesis pathway. The Clpex mutation decreases surface GPI expression. Surprisingly, Pgap2 showed tissue-specific expression with enrichment in the brain and face. We found the Clpex phenotype is due to apoptosis of neural crest cells (NCCs) and the cranial neuroepithelium. We showed folinic acid supplementation in utero can partially rescue the cleft lip phenotype. Finally, we generated a novel mouse model of NCC-specific total GPI deficiency. These mutants developed median cleft lip and palate demonstrating a previously undocumented cell autonomous role for GPI biosynthesis in NCC development.
Collapse
Affiliation(s)
- Marshall Lukacs
- Division of Human Genetics, Cincinnati Children's Medical Center, Cincinnati, United States.,Medical Scientist Training Program, Cincinnati Children's Medical Center, Cincinnati, United States
| | - Tia Roberts
- Division of Human Genetics, Cincinnati Children's Medical Center, Cincinnati, United States
| | - Praneet Chatuverdi
- Division of Developmental Biology, Cincinnati Children's Medical Center, Cincinnati, United States
| | - Rolf W Stottmann
- Division of Human Genetics, Cincinnati Children's Medical Center, Cincinnati, United States.,Medical Scientist Training Program, Cincinnati Children's Medical Center, Cincinnati, United States.,Division of Developmental Biology, Cincinnati Children's Medical Center, Cincinnati, United States.,Department of Pediatrics, University of Cincinnati, Cincinnati, United States
| |
Collapse
|
18
|
Saito-Diaz K, Zeltner N. Induced pluripotent stem cells for disease modeling, cell therapy and drug discovery in genetic autonomic disorders: a review. Clin Auton Res 2019; 29:367-384. [PMID: 30631982 DOI: 10.1007/s10286-018-00587-4] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2018] [Accepted: 12/26/2018] [Indexed: 12/19/2022]
Abstract
The autonomic nervous system (ANS) regulates all organs in the body independent of consciousness, and is thus essential for maintaining homeostasis of the entire organism. Diseases of the ANS can arise due to environmental insults such as injury, toxins/drugs and infections or due to genetic lesions. Human studies and animal models have been instrumental to understanding connectivity and regulation of the ANS and its disorders. However, research into cellular pathologies and molecular mechanisms of ANS disorders has been hampered by the difficulties in accessing human patient-derived ANS cells in large numbers to conduct meaningful research, mainly because patient neurons cannot be easily biopsied and primary human neuronal cultures cannot be expanded.Human-induced pluripotent stem cell (hiPSC) technology can elegantly bridge these issues, allowing unlimited access of patient-derived ANS cell types for cellular, molecular and biochemical analysis, facilitating the discovery of novel therapeutic targets, and eventually leading to drug discovery. Additionally, such cells may provide a source for cell replacement therapy to replenish lost or injured ANS tissue in patients.Here, we first review the anatomy and embryonic development of the ANS, as this knowledge is crucial for understanding disease modeling approaches. We then review the current advances in human stem cell technology for modeling diseases of the ANS, recent strides toward cell replacement therapy and drug discovery initiatives.
Collapse
Affiliation(s)
- Kenyi Saito-Diaz
- Center for Molecular Medicine, University of Georgia, Athens, GA, USA
| | - Nadja Zeltner
- Center for Molecular Medicine, University of Georgia, Athens, GA, USA. .,Department of Biochemistry and Molecular Biology, University of Georgia, Athens, GA, USA. .,Department of Cellular Biology, University of Georgia, Athens, GA, USA.
| |
Collapse
|
19
|
Wijeratne D, Rodger J, Stevenson A, Wallace H, Prêle CM, Wood FM, Fear MW. Ephrin-A2 affects wound healing and scarring in a murine model of excisional injury. Burns 2018; 45:682-690. [PMID: 30482614 DOI: 10.1016/j.burns.2018.10.002] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2018] [Revised: 09/14/2018] [Accepted: 10/04/2018] [Indexed: 12/18/2022]
Abstract
Ephrin ligand/Eph receptor signaling is important in both tissue development and homeostasis. There is increasing evidence that Ephrin/Eph signaling is important in the skin, involved in hair follicle cycling, epidermal differentiation, cutaneous innervation and skin cancer. However, there is currently limited information on the role of Ephrin/Eph signaling in cutaneous wound healing. Here we report the effects of the Ephrin-A2 and A5 ligands on wound healing. Using Ephrin-A2-/-, Ephrin-A5-/- and Ephrin-A2A5-/- transgenic mice, in vitro wound healing assays were conducted using isolated keratinocytes and fibroblasts. Ephrin-A2-/-, Ephrin-A2A5-/- and wild type mice with excisional wounds were used to analyze the impact of these ligands on wound closure, scar outcome, collagen orientation and re-innervation in vivo. The absence of the Ephrin-A2 and A5 ligands did not have any effect on dermal fibroblast proliferation or on fibroblast or keratinocyte migration. The loss of Ephrin-A2 and A5 ligands did not impact on the rate of wound closure or re-innervation after injury. However, changes in the gross morphology of the healed scar and in collagen histology of the scar dermis were observed in transgenic mice. Therefore Ephrin-A2 and A5 ligands may play an important role in final scar appearance associated with collagen deposition and structure.
Collapse
Affiliation(s)
- Dulharie Wijeratne
- Burn Injury Research Unit, School of Biomedical Sciences, The University of Western Australia, Australia
| | - Jennifer Rodger
- Experimental and Regenerative Neurosciences, School of Biological Sciences, The University of Western Australia, Australia
| | - Andrew Stevenson
- Burn Injury Research Unit, School of Biomedical Sciences, The University of Western Australia, Australia
| | - Hilary Wallace
- Burn Injury Research Unit, School of Biomedical Sciences, The University of Western Australia, Australia
| | - Cecilia M Prêle
- The Institute for Respiratory Health, The University of Western Australia, Nedlands, Western Australia, Australia; Centre for Cell Therapy and Regenerative Medicine, School of Biomedical Sciences, The University of Western Australia, Crawley, Western Australia, Australia
| | - Fiona M Wood
- Burn Injury Research Unit, School of Biomedical Sciences, The University of Western Australia, Australia; Centre for Cell Therapy and Regenerative Medicine, School of Biomedical Sciences, The University of Western Australia, Crawley, Western Australia, Australia; The Fiona Wood Foundation, Perth, Western Australia, Australia; Burns Service of Western Australia, WA Department of Health, Perth, Western Australia, Australia
| | - Mark W Fear
- Burn Injury Research Unit, School of Biomedical Sciences, The University of Western Australia, Australia; The Institute for Respiratory Health, The University of Western Australia, Nedlands, Western Australia, Australia; The Fiona Wood Foundation, Perth, Western Australia, Australia.
| |
Collapse
|
20
|
TBC1d24-ephrinB2 interaction regulates contact inhibition of locomotion in neural crest cell migration. Nat Commun 2018; 9:3491. [PMID: 30154457 PMCID: PMC6113226 DOI: 10.1038/s41467-018-05924-9] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2017] [Accepted: 08/02/2018] [Indexed: 11/08/2022] Open
Abstract
Although Eph-ephrin signalling has been implicated in the migration of cranial neural crest (CNC) cells, it is still unclear how ephrinB transduces signals regulating this event. We provide evidence that TBC1d24, a putative Rab35-GTPase activating protein (Rab35 GAP), complexes with ephrinB2 via the scaffold Dishevelled (Dsh) and mediates a signal affecting contact inhibition of locomotion (CIL) in CNC cells. Moreover, we found that, in migrating CNC, the interaction between ephrinB2 and TBC1d24 negatively regulates E-cadherin recycling in these cells via Rab35. Upon engagement of the cognate Eph receptor, ephrinB2 is tyrosine phosphorylated, which disrupts the ephrinB2/Dsh/TBC1d24 complex. The dissolution of this complex leads to increasing E-cadherin levels at the plasma membrane, resulting in loss of CIL and disrupted CNC migration. Our results indicate that TBC1d24 is a critical player in ephrinB2 control of CNC cell migration via CIL.
Collapse
|
21
|
Gouignard N, Andrieu C, Theveneau E. Neural crest delamination and migration: Looking forward to the next 150 years. Genesis 2018; 56:e23107. [PMID: 29675839 DOI: 10.1002/dvg.23107] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2018] [Revised: 03/27/2018] [Accepted: 03/28/2018] [Indexed: 12/24/2022]
Abstract
Neural crest (NC) cells were described for the first time in 1868 by Wilhelm His. Since then, this amazing population of migratory stem cells has been intensively studied. It took a century to fully unravel their incredible abilities to contribute to nearly every organ of the body. Yet, our understanding of the cell and molecular mechanisms controlling their migration is far from complete. In this review, we summarize the current knowledge on epithelial-mesenchymal transition and collective behavior of NC cells and propose further stops at which the NC train might be calling in the near future.
Collapse
Affiliation(s)
- Nadège Gouignard
- Centre de Biologie du Développement, Centre de Biologie Intégrative, Université de Toulouse, CNRS, UPS, France
| | - Cyril Andrieu
- Centre de Biologie du Développement, Centre de Biologie Intégrative, Université de Toulouse, CNRS, UPS, France
| | - Eric Theveneau
- Centre de Biologie du Développement, Centre de Biologie Intégrative, Université de Toulouse, CNRS, UPS, France
| |
Collapse
|
22
|
Okumura F, Joo-Okumura A, Obara K, Petersen A, Nishikimi A, Fukui Y, Nakatsukasa K, Kamura T. Ubiquitin ligase SPSB4 diminishes cell repulsive responses mediated by EphB2. Mol Biol Cell 2017; 28:3532-3541. [PMID: 28931592 PMCID: PMC5683763 DOI: 10.1091/mbc.e17-07-0450] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2017] [Revised: 09/08/2017] [Accepted: 09/13/2017] [Indexed: 11/29/2022] Open
Abstract
Eph receptor tyrosine kinases are important for cancer development and progression as well as in cellular repulsive responses. We determined that SOCS box-containing protein SPSB4 destabilizes EphB2 cytoplasmic fragments. SPSB4 is a novel ubiquitin ligase regulating EphB2-dependent cell repulsive responses. Eph receptor tyrosine kinases and their ephrin ligands are overexpressed in various human cancers, including colorectal malignancies, suggesting important roles in many aspects of cancer development and progression as well as in cellular repulsive responses. The ectodomain of EphB2 receptor is cleaved by metalloproteinases (MMPs) MMP-2/MMP-9 and released into the extracellular space after stimulation by its ligand. The remaining membrane-associated fragment is further cleaved by the presenilin-dependent γ-secretase and releases an intracellular peptide that has tyrosine kinase activity. Although the cytoplasmic fragment is degraded by the proteasome, the responsible ubiquitin ligase has not been identified. Here, we show that SOCS box-containing protein SPSB4 polyubiquitinates EphB2 cytoplasmic fragment and that SPSB4 knockdown stabilizes the cytoplasmic fragment. Importantly, SPSB4 down-regulation enhances cell repulsive responses mediated by EphB2 stimulation. Altogether, we propose that SPSB4 is a previously unidentified ubiquitin ligase regulating EphB2-dependent cell repulsive responses.
Collapse
Affiliation(s)
- Fumihiko Okumura
- Division of Biological Science, Graduate School of Science, Nagoya University, Aichi 464-8602, Japan
| | - Akiko Joo-Okumura
- Division of Biological Science, Graduate School of Science, Nagoya University, Aichi 464-8602, Japan
| | - Keisuke Obara
- Division of Biological Science, Graduate School of Science, Nagoya University, Aichi 464-8602, Japan
| | - Alexander Petersen
- Division of Biological Science, Graduate School of Science, Nagoya University, Aichi 464-8602, Japan
| | - Akihiko Nishikimi
- Division of Immunogenetics, Department of Immunobiology and Neuroscience, Medical Institute of Bioregulation, Kyushu University, Fukuoka 812-8582, Japan
| | - Yoshinori Fukui
- Division of Immunogenetics, Department of Immunobiology and Neuroscience, Medical Institute of Bioregulation, Kyushu University, Fukuoka 812-8582, Japan
| | - Kunio Nakatsukasa
- Division of Biological Science, Graduate School of Science, Nagoya University, Aichi 464-8602, Japan
| | - Takumi Kamura
- Division of Biological Science, Graduate School of Science, Nagoya University, Aichi 464-8602, Japan
| |
Collapse
|
23
|
Kemmerling N, Wunderlich P, Theil S, Linnartz-Gerlach B, Hersch N, Hoffmann B, Heneka MT, de Strooper B, Neumann H, Walter J. Intramembranous processing by γ-secretase regulates reverse signaling of ephrin-B2 in migration of microglia. Glia 2017; 65:1103-1118. [PMID: 28370426 DOI: 10.1002/glia.23147] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2015] [Revised: 03/14/2017] [Accepted: 03/16/2017] [Indexed: 12/30/2022]
Abstract
The Eph-ephrin system plays pivotal roles in cell adhesion and migration. The receptor-like functions of the ephrin ligands allow the regulation of intracellular processes via reverse signaling. γ-Secretase mediated processing of ephrin-B has previously been linked to activation of Src, a kinase crucial for focal adhesion and podosome phosphorylation. Here, we analyzed the role of γ-secretase in the stimulation of reverse ephrin-B2 signaling in the migration of mouse embryonic stem cell derived microglia. The proteolytic generation of the ephrin-B2 intracellular domain (ICD) by γ-secretase stimulates Src and focal adhesion kinase (FAK). Inhibition of γ-secretase decreased the phosphorylation of Src and FAK, and reduced cell motility. These effects were associated with enlargement of the podosomal surface. Interestingly, expression of ephrin-B2 ICD could rescue these effects, indicating that this proteolytic fragment mediates the activation of Src and FAK, and thereby regulates podosomal dynamics in microglial cells. Together, these results identify γ-secretase as well as ephrin-B2 as regulators of microglial migration.
Collapse
Affiliation(s)
- Nadja Kemmerling
- Department of Neurology, University of Bonn, Bonn, 53127, Germany
| | | | - Sandra Theil
- Department of Neurology, University of Bonn, Bonn, 53127, Germany
| | | | - Nils Hersch
- Institute of Complex Systems, ICS-7 Biomechanics, Forschungszentrum Jülich GmbH, Jülich, 52425, Germany
| | - Bernd Hoffmann
- Institute of Complex Systems, ICS-7 Biomechanics, Forschungszentrum Jülich GmbH, Jülich, 52425, Germany
| | - Michael T Heneka
- Department of Neurology, University of Bonn, Bonn, 53127, Germany.,German Center for Neurodegenerative Diseases, Bonn, 53127, Germany
| | - Bart de Strooper
- KULeuven Centre for Human Genetics, Leuven, 3000, Belgium.,Centre for Brain and Disease, VIB (Flanders Institute for Biotechnology), Leuven, 3000, Belgium
| | - Harald Neumann
- Institute of Reconstructive Neurobiology, University of Bonn, Bonn, 53127, Germany
| | - Jochen Walter
- Department of Neurology, University of Bonn, Bonn, 53127, Germany
| |
Collapse
|
24
|
Using approximate Bayesian computation to quantify cell-cell adhesion parameters in a cell migratory process. NPJ Syst Biol Appl 2017. [PMID: 28649436 PMCID: PMC5445583 DOI: 10.1038/s41540-017-0010-7] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
In this work, we implement approximate Bayesian computational methods to improve the design of a wound-healing assay used to quantify cell–cell interactions. This is important as cell–cell interactions, such as adhesion and repulsion, have been shown to play a role in cell migration. Initially, we demonstrate with a model of an unrealistic experiment that we are able to identify model parameters that describe agent motility and adhesion, given we choose appropriate summary statistics for our model data. Following this, we replace our model of an unrealistic experiment with a model representative of a practically realisable experiment. We demonstrate that, given the current (and commonly used) experimental set-up, our model parameters cannot be accurately identified using approximate Bayesian computation methods. We compare new experimental designs through simulation, and show more accurate identification of model parameters is possible by expanding the size of the domain upon which the experiment is performed, as opposed to increasing the number of experimental replicates. The results presented in this work, therefore, describe time and cost-saving alterations for a commonly performed experiment for identifying cell motility parameters. Moreover, this work will be of interest to those concerned with performing experiments that allow for the accurate identification of parameters governing cell migratory processes, especially cell migratory processes in which cell–cell adhesion or repulsion are known to play a significant role. Cell motility is a central process in wound healing and relies on complex cell-cell interactions. A team of mathematicians led by Ruth Baker and Kit Yates at the University of Oxford utilised computer simulations to re-design wound-healing assays that efficiently identify cell motility parameters. New experimental designs through computer simulation can more accurately identify cell motility parameters by expanding the size of the domain upon which the experiment is performed, as opposed to increasing the number of experimental replicates. The results describe time and cost-saving alterations for an experimental method for evaluate complex cell-cell interactions.
Collapse
|
25
|
Oiso N, Sakai K, Nishio K, Kawada A. Phylloid hypomelanosis associated with a mosaic trisomy 13 in the 13q31.3-qter region: atypical phylloid distribution and typical hypomelanosis. Pigment Cell Melanoma Res 2017; 30:269-272. [DOI: 10.1111/pcmr.12566] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Affiliation(s)
- Naoki Oiso
- Department of Dermatology; Kindai University Faculty of Medicine; Osaka-Sayama Japan
| | - Kazuko Sakai
- Department of Genome Biology; Kindai University Faculty of Medicine; Osaka-Sayama Japan
| | - Kazuto Nishio
- Department of Genome Biology; Kindai University Faculty of Medicine; Osaka-Sayama Japan
| | - Akira Kawada
- Department of Dermatology; Kindai University Faculty of Medicine; Osaka-Sayama Japan
| |
Collapse
|
26
|
Liu WC, Chen S, Zheng L, Qin L. Angiogenesis Assays for the Evaluation of Angiogenic Properties of Orthopaedic Biomaterials - A General Review. Adv Healthc Mater 2017; 6. [PMID: 28135051 DOI: 10.1002/adhm.201600434] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2016] [Revised: 06/04/2016] [Indexed: 01/07/2023]
Abstract
Vascularization is an essential process in bone formation, remodeling and regeneration during both bone development and fracture repair. Vascularization remains a big challenge directly leading to the final success of newly regenerated bone. In this review, the advantages and disadvantages of different angiogenesis assays and bone defect models are described in details for investigating revascularization of materials of interest. Unlike conventional angiogenesis study with growth factors or pharmaceutical molecules performed in two-dimension, special considerations are taken into account whether these assays can be translated for testing three-dimensional implantable devices. Over the years, accurate and quantifiable in vitro, ex vivo and in vivo assays have been extensively demonstrated to be useful in examining how new blood vessels grow. These methods can contribute to the fundamental understanding of angiogenic properties of the materials, but a bone defect model is still pivotal in order to understand the cascade actions of angiogenesis along with bone formation. Finally, angiogenesis and osteogenesis are both complex processes interacting with each other, the choice of which assay to be performed should adequately address the clinical relevance and reflect the sequence of responses of revascularization of the test materials.
Collapse
Affiliation(s)
- Wai Ching Liu
- Musculoskeletal Research Laboratory; Department of Orthopaedics & Traumatology; The Chinese University of Hong Kong; 5/F, Clinical Science Building, Prince of Wales Hospital Shatin Hong Kong SAR PR China
| | - Shihui Chen
- Musculoskeletal Research Laboratory; Department of Orthopaedics & Traumatology; The Chinese University of Hong Kong; 5/F, Clinical Science Building, Prince of Wales Hospital Shatin Hong Kong SAR PR China
- Pathology Center; Shanghai General Hospital/Faculty of Basic Medicine; Shanghai Jiao Tong University School of Medicine; Shanghai PR China
| | - Lizhen Zheng
- Musculoskeletal Research Laboratory; Department of Orthopaedics & Traumatology; The Chinese University of Hong Kong; 5/F, Clinical Science Building, Prince of Wales Hospital Shatin Hong Kong SAR PR China
| | - Ling Qin
- Musculoskeletal Research Laboratory; Department of Orthopaedics & Traumatology; The Chinese University of Hong Kong; 5/F, Clinical Science Building, Prince of Wales Hospital Shatin Hong Kong SAR PR China
- Translational Medicine R&D Center; Institute of Biomedical and Health Engineering; Shenzhen Institutes of Advanced Technology; Chinese Academy of Sciences; Shenzhen PR China
| |
Collapse
|
27
|
Hegde S, Srivastava O. Different gene knockout/transgenic mouse models manifesting persistent fetal vasculature: Are integrins to blame for this pathological condition? Life Sci 2016; 171:30-38. [PMID: 28039002 DOI: 10.1016/j.lfs.2016.12.019] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2016] [Revised: 12/15/2016] [Accepted: 12/26/2016] [Indexed: 12/30/2022]
Abstract
Persistent fetal vasculature (PFV) occurs as a result of a failure of fetal vasculature to undergo normal programmed involution. During development, before the formation of retinal vessels, the lens and the inner retina are nourished by the hyaloid vasculature. Hyaloid vessels extend from the optic nerve and run through the vitreous to encapsulate the lens. As fetal retinal vessels develop, hyaloid vasculature naturally regresses. Failure of regression of the hyaloid artery has been shown to lead to severe congenital pathologies. Studies on childhood blindness and visual impairment in the United States have shown that PFV accounts for 4.8% of total blindness. Although PFV is a serious developmental disease affecting the normal visual development pathway, the exact regulatory mechanism responsible for the regression of the hyaloid artery is still unknown. In this review, we have summarized the cellular defects associated with different knockout models that manifest features of persistent fetal vasculature. Based on similar cellular defects observed in different knockouts (KO)s such as altered migration, increased proliferation and decreased apoptosis and, the known role of integrins in the regulation of these cellular behaviors, we propose here that integrins may play a significant role in the pathophysiology of persistent fetal vasculature disease.
Collapse
Affiliation(s)
- Shylaja Hegde
- Department of Optometry and Vision Science, University of Alabama at Birmingham, Birmingham, AL 35294, United States.
| | - Om Srivastava
- Department of Optometry and Vision Science, University of Alabama at Birmingham, Birmingham, AL 35294, United States
| |
Collapse
|
28
|
O'Neill AK, Kindberg AA, Niethamer TK, Larson AR, Ho HYH, Greenberg ME, Bush JO. Unidirectional Eph/ephrin signaling creates a cortical actomyosin differential to drive cell segregation. J Cell Biol 2016; 215:217-229. [PMID: 27810913 PMCID: PMC5084648 DOI: 10.1083/jcb.201604097] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2016] [Accepted: 09/13/2016] [Indexed: 01/01/2023] Open
Abstract
Cell segregation is the process by which cells self-organize to establish developmental boundaries, an essential step in tissue formation. Cell segregation is a common outcome of Eph/ephrin signaling, but the mechanisms remain unclear. In craniofrontonasal syndrome, X-linked mosaicism for ephrin-B1 expression has been hypothesized to lead to aberrant Eph/ephrin-mediated cell segregation. Here, we use mouse genetics to exploit mosaicism to study cell segregation in the mammalian embryo and integrate live-cell imaging to examine the underlying cellular and molecular mechanisms. Our data demonstrate that dramatic ephrin-B1-mediated cell segregation occurs in the early neuroepithelium. In contrast to the paradigm that repulsive bidirectional signaling drives cell segregation, unidirectional EphB kinase signaling leads to cell sorting by the Rho kinase-dependent generation of a cortical actin differential between ephrin-B1- and EphB-expressing cells. These results define mechanisms of Eph/ephrin-mediated cell segregation, implicating unidirectional regulation of cortical actomyosin contractility as a key effector of this fundamental process.
Collapse
Affiliation(s)
- Audrey K O'Neill
- Department of Cell and Tissue Biology, Program in Craniofacial Biology and Institute for Human Genetics, University of California, San Francisco, San Francisco, CA 94143
| | - Abigail A Kindberg
- Department of Cell and Tissue Biology, Program in Craniofacial Biology and Institute for Human Genetics, University of California, San Francisco, San Francisco, CA 94143
- Biomedical Sciences Graduate Program, University of California, San Francisco, San Francisco, CA 94143
| | - Terren K Niethamer
- Department of Cell and Tissue Biology, Program in Craniofacial Biology and Institute for Human Genetics, University of California, San Francisco, San Francisco, CA 94143
- Biomedical Sciences Graduate Program, University of California, San Francisco, San Francisco, CA 94143
| | - Andrew R Larson
- Department of Cell and Tissue Biology, Program in Craniofacial Biology and Institute for Human Genetics, University of California, San Francisco, San Francisco, CA 94143
| | - Hsin-Yi Henry Ho
- Department of Cell Biology and Human Anatomy, University of California Davis School of Medicine, Davis, CA 95817
- Department of Neurobiology, Harvard Medical School, Boston, MA 02115
| | | | - Jeffrey O Bush
- Department of Cell and Tissue Biology, Program in Craniofacial Biology and Institute for Human Genetics, University of California, San Francisco, San Francisco, CA 94143
- Biomedical Sciences Graduate Program, University of California, San Francisco, San Francisco, CA 94143
- Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California, San Francisco, San Francisco, CA 94143
| |
Collapse
|
29
|
Menyhárt O, Harami-Papp H, Sukumar S, Schäfer R, Magnani L, de Barrios O, Győrffy B. Guidelines for the selection of functional assays to evaluate the hallmarks of cancer. Biochim Biophys Acta Rev Cancer 2016; 1866:300-319. [PMID: 27742530 DOI: 10.1016/j.bbcan.2016.10.002] [Citation(s) in RCA: 84] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2016] [Revised: 10/06/2016] [Accepted: 10/08/2016] [Indexed: 01/05/2023]
Abstract
The hallmarks of cancer capture the most essential phenotypic characteristics of malignant transformation and progression. Although numerous factors involved in this multi-step process are still unknown to date, an ever-increasing number of mutated/altered candidate genes are being identified within large-scale cancer genomic projects. Therefore, investigators need to be aware of available and appropriate techniques capable of determining characteristic features of each hallmark. We review the methods tailored to experimental cancer researchers to evaluate cell proliferation, programmed cell death, replicative immortality, induction of angiogenesis, invasion and metastasis, genome instability, and reprogramming of energy metabolism. Selecting the ideal method is based on the investigator's goals, available equipment and also on financial constraints. Multiplexing strategies enable a more in-depth data collection from a single experiment - obtaining several results from a single procedure reduces variability and saves time and relative cost, leading to more robust conclusions compared to a single end point measurement. Each hallmark possesses characteristics that can be analyzed by immunoblot, RT-PCR, immunocytochemistry, immunoprecipitation, RNA microarray or RNA-seq. In general, flow cytometry, fluorescence microscopy, and multiwell readers are extremely versatile tools and, with proper sample preparation, allow the detection of a vast number of hallmark features. Finally, we also provide a list of hallmark-specific genes to be measured in transcriptome-level studies. Although our list is not exhaustive, we provide a snapshot of the most widely used methods, with an emphasis on methods enabling the simultaneous evaluation of multiple hallmark features.
Collapse
Affiliation(s)
- Otília Menyhárt
- MTA TTK Lendület Cancer Biomarker Research Group, Magyar tudósok körútja 2, H-1117 Budapest, Hungary
| | | | - Saraswati Sukumar
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Reinhold Schäfer
- German Cancer Consortium (DKTK), DKFZ, Im Neuenheimer Feld 280, D-69120 Heidelberg and Charité Comprehensive Cancer Center, Invalidenstr. 80, D-10115 Berlin, Germany
| | - Luca Magnani
- Department of Surgery and Cancer, Imperial College London, London W12 0NN, UK
| | - Oriol de Barrios
- Group of Transcriptional Regulation of Gene Expression, Department of Oncology and Hematology, IDIBAPS, Barcelona, Spain
| | - Balázs Győrffy
- MTA TTK Lendület Cancer Biomarker Research Group, Magyar tudósok körútja 2, H-1117 Budapest, Hungary; 2nd Department of Pediatrics, Semmelweis University, H-1094 Budapest, Hungary.
| |
Collapse
|
30
|
Dixon KJ, Mier J, Gajavelli S, Turbic A, Bullock R, Turnley AM, Liebl DJ. EphrinB3 restricts endogenous neural stem cell migration after traumatic brain injury. Stem Cell Res 2016; 17:504-513. [PMID: 27771498 DOI: 10.1016/j.scr.2016.09.029] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/06/2016] [Revised: 08/29/2016] [Accepted: 09/24/2016] [Indexed: 10/20/2022] Open
Abstract
Traumatic brain injury (TBI) leads to a series of pathological events that can have profound influences on motor, sensory and cognitive functions. Conversely, TBI can also stimulate neural stem/progenitor cell proliferation leading to increased numbers of neuroblasts migrating outside their restrictive neurogenic zone to areas of damage in support of tissue integrity. Unfortunately, the factors that regulate migration are poorly understood. Here, we examine whether ephrinB3 functions to restrict neuroblasts from migrating outside the subventricular zone (SVZ) and rostral migratory stream (RMS). We have previously shown that ephrinB3 is expressed in tissues surrounding these regions, including the overlying corpus callosum (CC), and is reduced after controlled cortical impact (CCI) injury. Our current study takes advantage of ephrinB3 knockout mice to examine the influences of ephrinB3 on neuroblast migration into CC and cortex tissues after CCI injury. Both injury and/or ephrinB3 deficiency led to increased neuroblast numbers and enhanced migration outside the SVZ/RMS zones. Application of soluble ephrinB3-Fc molecules reduced neuroblast migration into the CC after injury and limited neuroblast chain migration in cultured SVZ explants. Our findings suggest that ephrinB3 expression in tissues surrounding neurogenic regions functions to restrict neuroblast migration outside the RMS by limiting chain migration.
Collapse
Affiliation(s)
- Kirsty J Dixon
- The Miami Project to Cure Paralysis, Department of Neurological Surgery, University of Miami, 1095 NW 14th Terrace, Miami, FL 33136, USA; Department of Physical Medicine and Rehabilitation, Virginia Commonwealth University, 1101 East Marshall Street, Richmond, VA 23298, USA.
| | - Jose Mier
- The Miami Project to Cure Paralysis, Department of Neurological Surgery, University of Miami, 1095 NW 14th Terrace, Miami, FL 33136, USA.
| | - Shyam Gajavelli
- The Miami Project to Cure Paralysis, Department of Neurological Surgery, University of Miami, 1095 NW 14th Terrace, Miami, FL 33136, USA.
| | - Alisa Turbic
- Department of Anatomy and Neuroscience, The University of Melbourne, Parkville, VIC 3010, Australia.
| | - Ross Bullock
- The Miami Project to Cure Paralysis, Department of Neurological Surgery, University of Miami, 1095 NW 14th Terrace, Miami, FL 33136, USA.
| | - Ann M Turnley
- Department of Anatomy and Neuroscience, The University of Melbourne, Parkville, VIC 3010, Australia.
| | - Daniel J Liebl
- The Miami Project to Cure Paralysis, Department of Neurological Surgery, University of Miami, 1095 NW 14th Terrace, Miami, FL 33136, USA.
| |
Collapse
|
31
|
Zhang S, Su Y, Gao J, Zhang C, Tanaka H. A potential inhibitory function of draxin in regulating mouse trunk neural crest migration. In Vitro Cell Dev Biol Anim 2016; 53:43-53. [PMID: 27649978 PMCID: PMC5258808 DOI: 10.1007/s11626-016-0079-0] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2016] [Accepted: 07/25/2016] [Indexed: 11/28/2022]
Abstract
Draxin is a repulsive axon guidance protein that plays important roles in the formation of three commissures in the central nervous system and dorsal interneuron 3 (dI3) in the chick spinal cord. In the present study, we report the expression pattern of mouse draxin in the embryonic mouse trunk spinal cord. In the presence of draxin, the longest net migration length of a migrating mouse trunk neural crest cell was significantly reduced. In addition, the relative number of apolar neural crest cells increased as the draxin treatment time increased. Draxin caused actin cytoskeleton rearrangement in the migrating trunk neural crest cells. Our data suggest that draxin may regulate mouse trunk neural crest cell migration by the rearrangement of cell actin cytoskeleton and by reducing the polarization activity of these cells subsequently.
Collapse
Affiliation(s)
- Sanbing Zhang
- Department of Hand and Foot Surgery, The Third Hospital of Shijiazhuang City, 15 South Tiyu Street, Shijiazhuang, 050000, People's Republic of China.
| | - Yuhong Su
- Department of Human Anatomy, Hebei Medical University, No. 361 East Zhongshan Road, Shijiazhuang, Hebei Province, 050017, People's Republic of China.
| | - Jinbao Gao
- Department of Hand and Foot Surgery, The Third Hospital of Shijiazhuang City, 15 South Tiyu Street, Shijiazhuang, 050000, People's Republic of China
| | - Chenbing Zhang
- Department of Hand and Foot Surgery, The Third Hospital of Shijiazhuang City, 15 South Tiyu Street, Shijiazhuang, 050000, People's Republic of China
| | - Hideaki Tanaka
- Division of Developmental Neurobiology, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| |
Collapse
|
32
|
George L, Dunkel H, Hunnicutt BJ, Filla M, Little C, Lansford R, Lefcort F. In vivo time-lapse imaging reveals extensive neural crest and endothelial cell interactions during neural crest migration and formation of the dorsal root and sympathetic ganglia. Dev Biol 2016; 413:70-85. [PMID: 26988118 PMCID: PMC4834247 DOI: 10.1016/j.ydbio.2016.02.028] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2015] [Revised: 02/11/2016] [Accepted: 02/27/2016] [Indexed: 11/21/2022]
Abstract
During amniote embryogenesis the nervous and vascular systems interact in a process that significantly affects the respective morphogenesis of each network by forming a "neurovascular" link. The importance of neurovascular cross-talk in the central nervous system has recently come into focus with the growing awareness that these two systems interact extensively both during development, in the stem-cell niche, and in neurodegenerative conditions such as Alzheimer's Disease and Amyotrophic Lateral Sclerosis. With respect to the peripheral nervous system, however, there have been no live, real-time investigations of the potential relationship between these two developing systems. To address this deficit, we used multispectral 4D time-lapse imaging in a transgenic quail model in which endothelial cells (ECs) express a yellow fluorescent marker, while neural crest cells (NCCs) express an electroporated red fluorescent marker. We monitored EC and NCC migration in real-time during formation of the peripheral nervous system. Our time-lapse recordings indicate that NCCs and ECs are physically juxtaposed and dynamically interact at multiple locations along their trajectories. These interactions are stereotypical and occur at precise anatomical locations along the NCC migratory pathway. NCCs migrate alongside the posterior surface of developing intersomitic vessels, but fail to cross these continuous streams of motile ECs. NCCs change their morphology and migration trajectory when they encounter gaps in the developing vasculature. Within the nascent dorsal root ganglion, proximity to ECs causes filopodial retraction which curtails forward persistence of NCC motility. Overall, our time-lapse recordings support the conclusion that primary vascular networks substantially influence the distribution and migratory behavior of NCCs and the patterned formation of dorsal root and sympathetic ganglia.
Collapse
Affiliation(s)
- Lynn George
- Department of Cell Biology and Neuroscience, Montana State University, Bozeman, MT 59717, United States; Department of Biological and Physical Sciences, Montana State University Billings, Billings, MT 59101, United States.
| | - Haley Dunkel
- Department of Cell Biology and Neuroscience, Montana State University, Bozeman, MT 59717, United States
| | - Barbara J Hunnicutt
- Department of Cell Biology and Neuroscience, Montana State University, Bozeman, MT 59717, United States
| | - Michael Filla
- University of Kansas Medical Center, Kansas City, KS 66160, United States
| | - Charles Little
- University of Kansas Medical Center, Kansas City, KS 66160, United States
| | - Rusty Lansford
- Department of Radiology and Developmental Neuroscience Program, Saban Research Institute, Children's Hospital Los Angeles, Los Angeles, CA 90027, United States; Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, United States
| | - Frances Lefcort
- Department of Cell Biology and Neuroscience, Montana State University, Bozeman, MT 59717, United States
| |
Collapse
|
33
|
Morrison MA, Zimmerman MW, Look AT, Stewart RA. Studying the peripheral sympathetic nervous system and neuroblastoma in zebrafish. Methods Cell Biol 2016; 134:97-138. [PMID: 27312492 DOI: 10.1016/bs.mcb.2015.12.003] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
The zebrafish serves as an excellent model to study vertebrate development and disease. Optically clear embryos, combined with tissue-specific fluorescent reporters, permit direct visualization and measurement of peripheral nervous system formation in real time. Additionally, the model is amenable to rapid cellular, molecular, and genetic approaches to determine how developmental mechanisms contribute to disease states, such as cancer. In this chapter, we describe the development of the peripheral sympathetic nervous system (PSNS) in general, and our current understanding of genetic pathways important in zebrafish PSNS development specifically. We also illustrate how zebrafish genetics is used to identify new mechanisms controlling PSNS development and methods for interrogating the potential role of PSNS developmental pathways in neuroblastoma pathogenesis in vivo using the zebrafish MYCN-driven neuroblastoma model.
Collapse
Affiliation(s)
- M A Morrison
- University of Utah, Salt Lake City, UT, United States
| | | | - A T Look
- Harvard Medical School, Boston, MA, United States
| | - R A Stewart
- University of Utah, Salt Lake City, UT, United States
| |
Collapse
|
34
|
Barsotti AM, Ryskin M, Zhong W, Zhang WG, Giannakou A, Loreth C, Diesl V, Follettie M, Golas J, Lee M, Nichols T, Fan C, Li G, Dann S, Fantin VR, Arndt K, Verhelle D, Rollins RA. Epigenetic reprogramming by tumor-derived EZH2 gain-of-function mutations promotes aggressive 3D cell morphologies and enhances melanoma tumor growth. Oncotarget 2015; 6:2928-38. [PMID: 25671303 PMCID: PMC4413628 DOI: 10.18632/oncotarget.2758] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2014] [Accepted: 11/16/2014] [Indexed: 12/20/2022] Open
Abstract
In addition to genetic alterations, cancer cells are characterized by myriad epigenetic changes. EZH2 is a histone methyltransferase that is over-expressed and mutated in cancer. The EZH2 gain-of-function (GOF) mutations first identified in lymphomas have recently been reported in melanoma (~2%) but remain uncharacterized. We expressed multiple EZH2 GOF mutations in the A375 metastatic skin melanoma cell line and observed both increased H3K27me3 and dramatic changes in 3D culture morphology. In these cells, prominent morphological changes were accompanied by a decrease in cell contractility and an increase in collective cell migration. At the molecular level, we observed significant alteration of the axonal guidance pathway, a pathway intricately involved in the regulation of cell shape and motility. Furthermore, the aggressive 3D morphology of EZH2 GOF-expressing melanoma cells (both endogenous and ectopic) was attenuated by EZH2 catalytic inhibition. Finally, A375 cells expressing exogenous EZH2 GOF mutants formed larger tumors than control cells in mouse xenograft studies. This study not only demonstrates the first functional characterization of EZH2 GOF mutants in non-hematopoietic cells, but also provides a rationale for EZH2 catalytic inhibition in melanoma.
Collapse
Affiliation(s)
- Anthony M Barsotti
- Oncology Research Unit, Pfizer Worldwide Research and Development, Pearl River, NY 10965, USA
| | - Michael Ryskin
- Oncology Research Unit, Pfizer Worldwide Research and Development, Pearl River, NY 10965, USA
| | - Wenyan Zhong
- Oncology Research Unit, Pfizer Worldwide Research and Development, Pearl River, NY 10965, USA
| | - Wei-Guo Zhang
- Oncology Research Unit, Pfizer Worldwide Research and Development, Pearl River, NY 10965, USA
| | - Andreas Giannakou
- Oncology Research Unit, Pfizer Worldwide Research and Development, Pearl River, NY 10965, USA
| | - Christine Loreth
- Oncology Research Unit, Pfizer Worldwide Research and Development, Cambridge, MA 02140, USA
| | - Veronica Diesl
- Oncology Research Unit, Pfizer Worldwide Research and Development, Cambridge, MA 02140, USA
| | - Maximillian Follettie
- Oncology Research Unit, Pfizer Worldwide Research and Development, Pearl River, NY 10965, USA.,Oncology Research Unit, Pfizer Worldwide Research and Development, Cambridge, MA 02140, USA
| | - Jonathon Golas
- Oncology Research Unit, Pfizer Worldwide Research and Development, Pearl River, NY 10965, USA
| | - Michelle Lee
- Drug Safety Research and Development, Pfizer Worldwide Research and Development, San Diego, CA 92121, USA
| | - Timothy Nichols
- Drug Safety Research and Development, Pfizer Worldwide Research and Development, San Diego, CA 92121, USA
| | - Conglin Fan
- Oncology Research Unit, Pfizer Worldwide Research and Development, San Diego, CA 92121, USA
| | - Gang Li
- Oncology Research Unit, Pfizer Worldwide Research and Development, San Diego, CA 92121, USA
| | - Stephen Dann
- Oncology Research Unit, Pfizer Worldwide Research and Development, San Diego, CA 92121, USA
| | - Valeria R Fantin
- Oncology Research Unit, Pfizer Worldwide Research and Development, San Diego, CA 92121, USA
| | - Kim Arndt
- Oncology Research Unit, Pfizer Worldwide Research and Development, Pearl River, NY 10965, USA
| | - Dominique Verhelle
- Oncology Research Unit, Pfizer Worldwide Research and Development, San Diego, CA 92121, USA
| | - Robert A Rollins
- Oncology Research Unit, Pfizer Worldwide Research and Development, Pearl River, NY 10965, USA
| |
Collapse
|
35
|
Nunan R, Campbell J, Mori R, Pitulescu ME, Jiang WG, Harding KG, Adams RH, Nobes CD, Martin P. Ephrin-Bs Drive Junctional Downregulation and Actin Stress Fiber Disassembly to Enable Wound Re-epithelialization. Cell Rep 2015; 13:1380-1395. [PMID: 26549443 PMCID: PMC4660216 DOI: 10.1016/j.celrep.2015.09.085] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2015] [Revised: 08/12/2015] [Accepted: 09/30/2015] [Indexed: 12/21/2022] Open
Abstract
For a skin wound to successfully heal, the cut epidermal-edge cells have to migrate forward at the interface between scab and healthy granulation tissue. Much is known about how lead-edge cells migrate, but very little is known about the mechanisms that enable active participation by cells further back. Here we show that ephrin-B1 and its receptor EphB2 are both upregulated in vivo, just for the duration of repair, in the first 70 or so rows of epidermal cells, and this signal leads to downregulation of the molecular components of adherens and tight (but not desmosomal) junctions, leading to loosening between neighbors and enabling shuffle room among epidermal cells. Additionally, this signaling leads to the shutdown of actomyosin stress fibers in these same epidermal cells, which may act to release tension within the wound monolayer. If this signaling axis is perturbed, then disrupted healing is a consequence in mouse and man. Ephrin-B/EphBs are upregulated in the migrating wound epidermis in mouse and man Ephrin-B/EphB signaling drives junction loosening, thus enabling re-epithelialization Ephrin-B/EphB signaling also leads to dissolution of stress fibers and tension release In human chronic wounds ephrin-Bs are misregulated and may be a therapeutic target
Collapse
Affiliation(s)
- Robert Nunan
- Schools of Biochemistry and Physiology & Pharmacology, University of Bristol, Bristol BS8 1TD, UK
| | - Jessica Campbell
- Schools of Biochemistry and Physiology & Pharmacology, University of Bristol, Bristol BS8 1TD, UK
| | - Ryoichi Mori
- Schools of Biochemistry and Physiology & Pharmacology, University of Bristol, Bristol BS8 1TD, UK; Department of Pathology, Nagasaki University, Nagasaki 852-8523, Japan
| | - Mara E Pitulescu
- Max Planck Institute for Molecular Biomedicine, 48149 Muenster, Germany; Faculty of Medicine, University of Muenster, 48149 Muenster, Germany
| | - Wen G Jiang
- School of Medicine, Cardiff University, Cardiff CF14 4XN, UK
| | - Keith G Harding
- School of Medicine, Cardiff University, Cardiff CF14 4XN, UK
| | - Ralf H Adams
- Max Planck Institute for Molecular Biomedicine, 48149 Muenster, Germany; Faculty of Medicine, University of Muenster, 48149 Muenster, Germany
| | - Catherine D Nobes
- Schools of Biochemistry and Physiology & Pharmacology, University of Bristol, Bristol BS8 1TD, UK
| | - Paul Martin
- Schools of Biochemistry and Physiology & Pharmacology, University of Bristol, Bristol BS8 1TD, UK; School of Medicine, Cardiff University, Cardiff CF14 4XN, UK.
| |
Collapse
|
36
|
Ross RJ, Yates C, Baker R. Inference of cell–cell interactions from population density characteristics and cell trajectories on static and growing domains. Math Biosci 2015; 264:108-18. [DOI: 10.1016/j.mbs.2015.04.002] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2014] [Revised: 03/31/2015] [Accepted: 04/02/2015] [Indexed: 01/17/2023]
|
37
|
Myers JN, Davis L, Sheehan D, Kulharya AS. Mosaic tetrasomy 13q and phylloid hypomelanosis: a case report and review of the literature. Pediatr Dermatol 2015; 32:263-6. [PMID: 24920397 DOI: 10.1111/pde.12375] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
A 6-year-old girl presented for evaluation of skin discoloration. Examination revealed oval and oblong hypopigmented macules on her trunk and extremities. Cytogenetic studies and immunohistochemistry of biopsies from normally pigmented and hypopigmented skin revealed mosaicism for partial tetrasomy for 13q with low melanocyte levels in lesional skin. The patient was diagnosed with phylloid hypomelanosis (PH), a distinct clinical entity linked to abnormalities in chromosome 13. This article reviews the literature regarding PH and supports the notion that mosaicism of the melanocyte region of chromosome 13q is responsible for PH.
Collapse
Affiliation(s)
- Joseph N Myers
- Division of Dermatology, Georgia Regents University, Augusta, Georgia
| | | | | | | |
Collapse
|
38
|
Park I, Lee HS. EphB/ephrinB signaling in cell adhesion and migration. Mol Cells 2015; 38:14-9. [PMID: 25475547 PMCID: PMC4314128 DOI: 10.14348/molcells.2015.2116] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2014] [Revised: 10/14/2014] [Accepted: 10/17/2014] [Indexed: 11/27/2022] Open
Abstract
Eph receptors and their ligands, ephrins, represent the largest group of the receptor tyrosine kinase (RTK) family, and they mediate numerous developmental processes in a variety of organisms. Ephrins are membrane-bound proteins that are mainly divided into two classes: A class ephrins, which are linked to the membrane by a glycosylphosphatidylinositol (GPI) linkage, and B class ephrins, which are transmembrane ligands. Based on their domain structures and affinities for ligand binding, the Eph receptors are also divided into two groups. Trans-dimerization of Eph receptors with their membrane-tethered ligands regulates cell-cell interactions and initiates bidirectional signaling pathways. These pathways are intimately involved in regulating cytoskeleton dynamics, cell migration, and alterations in cellular dynamics and shapes. The EphBs and ephrinBs are specifically localized and modified to promote higher-order clustering and initiate of bidirectional signaling. In this review, we present an in-depth overview of the structure, mechanisms, cell signaling, and functions of EphB/ephrinB in cell adhesion and migration.
Collapse
Affiliation(s)
- Inji Park
- ABRC, CMRI, School of Life Sciences, BK21 Plus KNU Creative BioResearch Group, Kyungpook National University, Daegu 702-701,
Korea
| | - Hyun-Shik Lee
- ABRC, CMRI, School of Life Sciences, BK21 Plus KNU Creative BioResearch Group, Kyungpook National University, Daegu 702-701,
Korea
| |
Collapse
|
39
|
Maguire LH, Thomas AR, Goldstein AM. Tumors of the neural crest: Common themes in development and cancer. Dev Dyn 2014; 244:311-22. [PMID: 25382669 DOI: 10.1002/dvdy.24226] [Citation(s) in RCA: 79] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2014] [Revised: 10/31/2014] [Accepted: 11/03/2014] [Indexed: 12/17/2022] Open
Abstract
The neural crest (NC) is a remarkable transient structure in the vertebrate embryo that gives rise to a highly versatile population of pluripotent cells that contribute to the formation of multiple tissues and organs throughout the body. In order to achieve their task, NC-derived cells have developed specialized mechanisms to promote (1) their transition from an epithelial to a mesenchymal phenotype, (2) their capacity for extensive migration and cell proliferation, and (3) their ability to produce diverse cell types largely depending on the microenvironment encountered during and after their migratory path. Following embryogenesis, these same features of cellular motility, invasion, and proliferation can become a liability by contributing to tumorigenesis and metastasis. Ample evidence has shown that cancer cells have cleverly co-opted many of the genetic and molecular features used by developing NC cells. This review focuses on tumors that arise from NC-derived tissues and examines mechanistic themes shared during their oncogenic and metastatic development with embryonic NC cell ontogeny.
Collapse
Affiliation(s)
- Lillias H Maguire
- Department of Pediatric Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | | | | |
Collapse
|
40
|
Fort P, Théveneau E. PleiotRHOpic: Rho pathways are essential for all stages of Neural Crest development. Small GTPases 2014; 5:e27975. [PMID: 24614304 DOI: 10.4161/sgtp.27975] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Neural Crest (NC) cells are a multipotent migratory stem cell population unique to vertebrates, which contributes extensively to the formation of a wide array of neural and non-neural structures in the embryo. NC cells originate in the ectoderm at the border of the neural tube, undergo an epithelial-mesenchymal transition and acquire outstanding individual and collective migratory properties that allow them to disseminate and differentiate to different parts of the body. This exquisite capacity to switch from an epithelium to motile cells represents both a puzzling biological issue and an attractive model to address the basic mechanisms of cell migration and their alteration during cancer progression. Here we review how signaling pathways controlled by Rho GTPases, key players in cell adhesion, contraction, migration and polarity, contribute to the control the different phases of NC development.
Collapse
Affiliation(s)
- Philippe Fort
- CNRS; University Montpellier 2; CRBM-UMR5237; Montpellier, France
| | - Eric Théveneau
- CNRS; University Toulouse III; Centre de Biologie du Développement; UMR5547; Toulouse, France
| |
Collapse
|
41
|
The role of the non-canonical Wnt-planar cell polarity pathway in neural crest migration. Biochem J 2014; 457:19-26. [PMID: 24325550 DOI: 10.1042/bj20131182] [Citation(s) in RCA: 77] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
The neural crest is an embryonic stem cell population whose migratory behaviour has been likened to malignant invasion. The neural crest, as does cancer, undergoes an epithelial-to-mesenchymal transition and migrates to colonize almost all the tissues of the embryo. Neural crest cells exhibit collective cell migration, moving in streams of high directionality. The migratory neural crest streams are kept in shape by the presence of negative signals in their vicinity. The directionality of the migrating neural crest is achieved by contact-dependent cell polarization, in a phenomenon called contact inhibition of locomotion. Two cells experiencing contact inhibition of locomotion move away from each other after collision. However, if the cell density is high only cells exposed to a free edge can migrate away from the cluster leading to the directional migration of the whole group. Recent work performed in chicks, zebrafish and frogs has shown that the non-canonical Wnt-PCP (planar cell polarity) pathway plays a major role in neural crest migration. PCP signalling controls contact inhibition of locomotion between neural crest cells by localizing different PCP proteins at the site of cell contact during collision and locally regulating the activity of Rho GTPases. Upon collision RhoA (ras homologue family member A) is activated, whereas Rac1 is inhibited at the contact between two migrating neural crest cells, leading to the collapse of protrusions and the migration of cells away from one another. The present review summarizes the mechanisms that control neural crest migration and focuses on the role of non-canonical Wnt or PCP signalling in this process.
Collapse
|
42
|
Steinecke A, Gampe C, Zimmer G, Rudolph J, Bolz J. EphA/ephrin A reverse signaling promotes the migration of cortical interneurons from the medial ganglionic eminence. Development 2014; 141:460-71. [DOI: 10.1242/dev.101691] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Inhibitory interneurons control the flow of information and synchronization in the cerebral cortex at the circuit level. During embryonic development, multiple subtypes of cortical interneurons are generated in different regions of the ventral telencephalon, such as the medial and caudal ganglionic eminence (MGE and CGE), as well as the preoptic area (POA). These neurons then migrate over long distances towards their cortical target areas. Diverse families of diffusible and cell-bound signaling molecules, including the Eph/ephrin system, regulate and orchestrate interneuron migration. Ephrin A3 and A5, for instance, are expressed at the borders of the pathway of MGE-derived interneurons and prevent these cells from entering inappropriate regions via EphA4 forward signaling. We found that MGE-derived interneurons, in addition to EphA4, also express ephrin A and B ligands, suggesting Eph/ephrin forward and reverse signaling in the same cell. In vitro and in vivo approaches showed that EphA4-induced reverse signaling in MGE-derived interneurons promotes their migration and that this effect is mediated by ephrin A2 ligands. In EphA4 mutant mice, as well as after ephrin A2 knockdown using in utero electroporation, we found delayed interneuron migration at embryonic stages. Thus, besides functions in guiding MGE-derived interneurons to the cortex through forward signaling, here we describe a novel role of the ephrins in driving these neurons to their target via reverse signaling.
Collapse
Affiliation(s)
- André Steinecke
- Universität Jena, Institut für Allgemeine Zoologie und Tierphysiologie, 07743 Jena, Germany
| | - Christin Gampe
- Universität Jena, Institut für Allgemeine Zoologie und Tierphysiologie, 07743 Jena, Germany
| | - Geraldine Zimmer
- Universität Jena, Institut für Allgemeine Zoologie und Tierphysiologie, 07743 Jena, Germany
| | - Judith Rudolph
- Universität Jena, Institut für Allgemeine Zoologie und Tierphysiologie, 07743 Jena, Germany
| | - Jürgen Bolz
- Universität Jena, Institut für Allgemeine Zoologie und Tierphysiologie, 07743 Jena, Germany
| |
Collapse
|
43
|
Freitas PF, Borges ME, Ortolani-Machado CF. The distribution of ephrin-B1 and PNA-positive glycoconjugates is correlated with atypical melanoblast migration in Japanese Silky fowl embryos. Acta Histochem 2014; 116:138-47. [PMID: 23916864 DOI: 10.1016/j.acthis.2013.06.006] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2013] [Revised: 06/24/2013] [Accepted: 06/25/2013] [Indexed: 10/26/2022]
Abstract
Melanoblasts are positively stimulated to migrate in the dorsolateral pathway of the avian embryo by ephrins, but are inhibited by PNA-binding glycoconjugates. We analyzed the potential role of these molecules in the Japanese Silky fowl, which displays intense internal pigmentation. The distribution of ephrin ligands was analyzed using Eph receptor-human Fc fusion proteins. Glycoconjugates were labeled using PNA-FITC. In Japanese Silky embryos, ventral areas, including the anterior- and posterior-half somites, expressed ephrin-B1 in a pattern that correlates with the atypical migratory pathways taken by Japanese Silky melanoblasts. White Leghorn embryos displayed little to no ephrin-Bs in the ventral paths. Conversely, PNA-binding barrier tissues, proposed to prevent melanoblasts from migrating ventrally in White Leghorn, are missing or have significant gaps in Japanese Silky embryos. Thus, studies of a naturally occurring pigmentation mutant confirm that a combination of cues regulates melanoblast migration in the chick embryo.
Collapse
|
44
|
Sivka U, Snoj A, Palandačić A, Sušnik Bajec S. Identification of candidate genes involved in marble color pattern formation in genus Salmo. COMPARATIVE BIOCHEMISTRY AND PHYSIOLOGY D-GENOMICS & PROTEOMICS 2013; 8:244-9. [PMID: 23850874 DOI: 10.1016/j.cbd.2013.06.003] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/11/2013] [Revised: 06/21/2013] [Accepted: 06/21/2013] [Indexed: 10/26/2022]
Abstract
Salmonids are known for their variety in skin color and color patterning, which depends on reaction-diffusion mechanism or/and cell-cell interaction. One of the visually most prominent characteristics found in the genus Salmo is the marble color pattern, distinctive for marble trout (Salmo marmoratus). In order to identify and characterize genes potentially involved in skin coloration and color pattern formation in marble trout, a salmonid 32K cDNA microarray was applied to compare skin transcriptome profiles from marble trout, brown trout (Salmo trutta), and marble trout×brown trout hybrids exhibiting the marble color pattern. Microarray results were validated by qRT-PCR and revealed four differentially expressed informative genes (hdac1, vps18, dct and scg2a) involved in animal pigmentation. hdac1 and dct were associated with the wnt signaling pathway, vps18 was involved in melanosome biogenesis and scg2a was observed to act as physiological factor in skin pigmentation and thus could be regarded as a helper protein in prohormone Pomc packaging. We propose that the formation of the marble color pattern is at least partially based upon a reaction-diffusion mechanism and depends upon wnt signaling pathway.
Collapse
Affiliation(s)
- U Sivka
- Biotechnical Faculty, Department of Animal Science, University of Ljubljana, Groblje 3, SI-1230 Domžale, Slovenia.
| | | | | | | |
Collapse
|
45
|
Teslaa JJ, Keller AN, Nyholm MK, Grinblat Y. Zebrafish Zic2a and Zic2b regulate neural crest and craniofacial development. Dev Biol 2013; 380:73-86. [PMID: 23665173 DOI: 10.1016/j.ydbio.2013.04.033] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2012] [Revised: 04/25/2013] [Accepted: 04/29/2013] [Indexed: 11/25/2022]
Abstract
Holoprosencephaly (HPE), the most common malformation of the human forebrain, is associated with defects of the craniofacial skeleton. ZIC2, a zinc-finger transcription factor, is strongly linked to HPE and to a characteristic set of dysmorphic facial features in humans. We have previously identified important functions for zebrafish Zic2 in the developing forebrain. Here, we demonstrate that ZIC2 orthologs zic2a and zic2b also regulate the forming zebrafish craniofacial skeleton, including the jaw and neurocranial cartilages, and use the zebrafish to study Zic2-regulated processes that may contribute to the complex etiology of HPE. Using temporally controlled Zic2a overexpression, we show that the developing craniofacial cartilages are sensitive to Zic2 elevation prior to 24hpf. This window of sensitivity overlaps the critical expansion and migration of the neural crest (NC) cells, which migrate from the developing neural tube to populate vertebrate craniofacial structures. We demonstrate that zic2b influences the induction of NC at the neural plate border, while both zic2a and zic2b regulate NC migratory onset and strongly contribute to chromatophore development. Both Zic2 depletion and early ectopic Zic2 expression cause moderate, incompletely penetrant mispatterning of the NC-derived jaw precursors at 24hpf, yet by 2dpf these changes in Zic2 expression result in profoundly mispatterned chondrogenic condensations. We attribute this discrepancy to an additional role for Zic2a and Zic2b in patterning the forebrain primordium, an important signaling source during craniofacial development. This hypothesis is supported by evidence that transplanted Zic2-deficient cells can contribute to craniofacial cartilages in a wild-type background. Collectively, these data suggest that zebrafish Zic2 plays a dual role during craniofacial development, contributing to two disparate aspects of craniofacial morphogenesis: (1) neural crest induction and migration, and (2) early patterning of tissues adjacent to craniofacial chondrogenic condensations.
Collapse
Affiliation(s)
- Jessica J Teslaa
- Department of Zoology, University of Wisconsin, Madison, WI 53706, USA
| | | | | | | |
Collapse
|
46
|
Domingues MJ, Larue L, Bonaventure J. [Migration of melanocytic lineage-derived cells]. Med Sci (Paris) 2013; 29:287-92. [PMID: 23544383 DOI: 10.1051/medsci/2013293015] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
During development, neural crest cells-derived melanoblasts migrate along the dorso-lateral axis into the dermis, then cross the basal layer to reach the epidermis and differentiate into melanocytes. They finally colonize the hair follicles to become resident pigmented cells. Neoplastic transformation converts melanocytes into highly invasive melanoma cells, which can adopt two modes of interconvertible migration (mesenchymal and amoeboid). Through analysis of the coat color phenotype of natural mouse mutants and genetically modified animals, many of the genes regulating migration were identified. Deciphering of cell membrane protrusions and signaling molecules involved in melanoma cell motility was further achieved through 2D and 3D culture systems. Here, we summarize how these data allow a better understanding of the complex mechanisms controlling migration of normal and pathological cells of the melanocytic lineage.
Collapse
Affiliation(s)
- Mélanie J Domingues
- Institut Curie, CNRS UMR3347, Inserm U1021,Génétique du développement des mélanocytes, Centre de recherche, bâtiment 110, 91405 Orsay, France
| | | | | |
Collapse
|
47
|
Bonaventure J, Domingues MJ, Larue L. Cellular and molecular mechanisms controlling the migration of melanocytes and melanoma cells. Pigment Cell Melanoma Res 2013; 26:316-25. [PMID: 23433358 DOI: 10.1111/pcmr.12080] [Citation(s) in RCA: 87] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2012] [Accepted: 02/07/2013] [Indexed: 10/27/2022]
Abstract
During embryonic development in vertebrates, the neural crest-derived melanoblasts migrate along the dorsolateral axis and cross the basal membrane separating the dermis from the epidermis to reach their final location in the interfollicular epidermis and epidermal hair follicles. Neoplastic transformation converts melanocytes into highly invasive and metastatic melanoma cells. In vitro, these cells extend various types of protrusions and adopt two interconvertible modes of migration, mesenchymal and amoeboid, driven by different signalling molecules. In this review, we describe the major contributions of natural mouse mutants, mouse models generated by genetic engineering and in vitro culture systems, to identification of the genes, signalling pathways and mechanisms regulating the migration of normal and pathological cells of the melanocyte lineage, at both the cellular and molecular levels.
Collapse
Affiliation(s)
- Jacky Bonaventure
- Developmental Genetics of Melanocytes, Institut Curie, Centre de Recherche, Orsay, France
| | | | | |
Collapse
|
48
|
Banerjee S, Isaacman-Beck J, Schneider VA, Granato M. A novel role for Lh3 dependent ECM modifications during neural crest cell migration in zebrafish. PLoS One 2013; 8:e54609. [PMID: 23349938 PMCID: PMC3548841 DOI: 10.1371/journal.pone.0054609] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2012] [Accepted: 12/14/2012] [Indexed: 12/11/2022] Open
Abstract
During vertebrate development, trunk neural crest cells delaminate along the entire length of the dorsal neural tube and initially migrate as a non-segmented sheet. As they enter the somites, neural crest cells rearrange into spatially restricted segmental streams. Extracellular matrix components are likely to play critical roles in this transition from a sheet-like to a stream-like mode of migration, yet the extracellular matrix components and their modifying enzymes critical for this transition are largely unknown. Here, we identified the glycosyltransferase Lh3, known to modify extracellular matrix components, and its presumptive substrate Collagen18A1, to provide extrinsic signals critical for neural crest cells to transition from a sheet-like migration behavior to migrating as a segmental stream. Using live cell imaging we show that in lh3 null mutants, neural crest cells fail to transition from a sheet to a stream, and that they consequently enter the somites as multiple streams, or stall shortly after entering the somites. Moreover, we demonstrate that transgenic expression of lh3 in a small subset of somitic cells adjacent to where neural crest cells switch from sheet to stream migration restores segmental neural crest cell migration. Finally, we show that knockdown of the presumptive Lh3 substrate Collagen18A1 recapitulates the neural crest cell migration defects observed in lh3 mutants, consistent with the notion that Lh3 exerts its effect on neural crest cell migration by regulating post-translational modifications of Collagen18A1. Together these data suggest that Lh3–Collagen18A1 dependent ECM modifications regulate the transition of trunk neural crest cells from a non-segmental sheet like migration mode to a segmental stream migration mode.
Collapse
Affiliation(s)
- Santanu Banerjee
- Department of Cell and Developmental Biology, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, United States of America
| | - Jesse Isaacman-Beck
- Department of Cell and Developmental Biology, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, United States of America
| | - Valerie A. Schneider
- Department of Cell and Developmental Biology, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, United States of America
| | - Michael Granato
- Department of Cell and Developmental Biology, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, United States of America
- * E-mail:
| |
Collapse
|
49
|
Dhanasobhon D, Savier E, Lelievre V. To phosphorylate or not to phosphorylate: Selective alterations in tyrosine kinase-inhibited EphB mutant mice. Cell Adh Migr 2013; 8:1-4. [PMID: 24589619 DOI: 10.4161/cam.27478] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
EphB tyrosine kinase receptors have been implicated in multiple developmental processes; however, the signaling mechanism underlying these events remains unclear. Through a triple knock-in mouse line for three neurally expressed EphBs, Sokis et al. demonstrated that EphB tyrosine kinase activity is required for axon guidance but does not influence synapse formation. This short communication highlights their study and appealing molecular approach that elucidated the functions of EphB tyrosine kinase during developmental events.
Collapse
Affiliation(s)
- Dhanasak Dhanasobhon
- Joint master in Neuroscience; University of Strasbourg-France; Strasbourg, France
| | - Elise Savier
- Joint master in Neuroscience; University of Strasbourg-France; Strasbourg, France
| | - Vincent Lelievre
- Joint master in Neuroscience; University of Strasbourg-France; Strasbourg, France
| |
Collapse
|
50
|
Sarnat HB, Flores-Sarnat L. Genetics of neural crest and neurocutaneous syndromes. HANDBOOK OF CLINICAL NEUROLOGY 2013; 111:309-14. [PMID: 23622181 DOI: 10.1016/b978-0-444-52891-9.00036-1] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
Neural crest progenitor cells are identified at the lateral margins of the neural placode at the time of gastrulation. With folding of the placode, these precursors are brought to the dorsal midline of the neural tube at the site of closure, become committed to neural crest lineage and almost immediately migrate peripherally to various predetermined sites in the body and then differentiate as a variety of cellular types in all three of the traditional "germ layers." All of these processes of migration and differentiation of neural crest are precisely genetically programed, temporally and spatially, by a variety of genes. Primary neurocutaneous syndromes are all very different diseases with different genetic mutations, but the unifying factor amongst them is that all are neurocristopathies and can be explained as such, including the tumor-suppressor function of several of these genes, especially those of neurofibromatosis 1 and 2 and tuberous sclerosis. This chapter reviews the principal genes that program neural crest development and also are documented, implicated, or suspected in the pathogenesis of neurocutaneous syndromes. Recent genetic discoveries are noted in epidermal nevus syndrome, including Proteus syndrome and their association with hemimegalencephaly and congenital infiltrating lipomatosis of the face.
Collapse
Affiliation(s)
- Harvey B Sarnat
- Departments of Clinical Neurosciences and Paediatrics, Division of Paediatric Neurology, University of Calgary, Alberta Children's Hospital, Calgary, Canada
| | | |
Collapse
|